Novel Semisynthetic Derivative of Antibiotic Eremomycin Active

Anacor Pharmaceuticals, Inc., 1060 East Meadow Circle, Palo Alto, California 94303, Galileo Pharmaceuticals, Inc., 5301 Patrick Henry,. DriVe Santa Cl...
0 downloads 0 Views 69KB Size
J. Med. Chem. 2007, 50, 3681-3685

3681

Novel Semisynthetic Derivative of Antibiotic Eremomycin Active against Drug-Resistant Gram-Positive Pathogens Including Bacillus anthracis† Kirk R. Maples,‡ Conrad Wheeler,‡ Emily Ip,‡ Jacob J. Plattner,‡ Daniel Chu,§ Yong-Kang Zhang,‡ Maria N. Preobrazhenskaya,*,| Svetlana S. Printsevskaya,| Svetlana E. Solovieva,| Evgenia N. Olsufyeva,| Henry Heine,⊥ Julie Lovchik,# and C. Richard Lyons# Anacor Pharmaceuticals, Inc., 1060 East Meadow Circle, Palo Alto, California 94303, Galileo Pharmaceuticals, Inc., 5301 Patrick Henry, DriVe Santa Clara, California 95054, Gause Institute of New Antibiotics, Bolshaya PirogoVskaya 11, Moscow 119021, Russia, USAMRIID Bacteriology DiVision, 1425 Porter Street, Fort Detrick, Frederick, Maryland 21702, and UniVersity of New Mexico Health Sciences Center, BMSB G41, MSC10 5550, 1 UniVersity of New Mexico, Albuquerque, New Mexico, 87131 ReceiVed January 3, 2007

Five adamantyl-containing carboxamides of eremomycin or vancomycin were synthesized and their antibacterial activities against some Gram-positive clinical isolates were investigated in vitro and in vivo. The adamantyl-2 amide of glycopeptide antibiotic eremomycin (1a in Chart 1, AN0900) was the most active compound and showed high activity against several Gram-positive pathogens: vancomycin-susceptible staphylococci and enterococci, glycopeptide-intermediate-resistant Staphylococcus aureus, and glycopeptideresistant enterococci. Compound 1a was equally active in vitro against both Ciprofloxacin-susceptible and -resistant Bacillus anthracis strains (MICs 0.25-0.5 µg/mL). It was distinguished by having a 2.8 h halflife (t1/2) in mice and a volume of distribution of 2.18 L/kg. Compound 1a was active against Staphylococcus aureus in mice (iv) and provided complete protection against a lethal intravenous challenge with vegetative B. anthracis bacilli and also in a murine pulmonary anthrax model in which mice were challenged with Bacillus anthracis spores. Introduction The discovery and development of new antibiotics is of crucial importance because our current arsenal of antibiotics is rapidly becoming obsolete. This is due, in large measure, to the spread of drug resistant pathogens that have evolved mechanisms to resist almost all available antibiotics. Furthermore, ease of transportation is creating a “global village” that can result in the potential for the devastating spread of infectious agents. Other bacterial pathogens are being stockpiled by terrorist groups and countries, which increases the threat of biological warfare. The mailing of letters containing B. anthracis spores to government officials and members of the media in the fall of 2001 emphasized the need for developing effective countermeasures against potential biowarfare agents such as anthrax. Anthrax is a zoonotic disease caused by the spore forming organism B. anthracis and occurs worldwide. The disease can occur in three forms: cutaneous, gastrointestinal, and inhalation. In spite of supportive care, including appropriate antibiotics, inhalation anthrax mortality is very high.1 The current recommended antibiotic therapy for B. anthracis exposure is ciprofloxacin or doxycycline, and these agents are indeed effective against most strains of B. anthracis.2 The ability to create resistant strains of B. anthracis, as demonstrated by Athamna et al.,3 resistant to ciprofloxacin and 17 other antibiotics, including the antibiotic vancomycin (2, Chart 1), is of concern. Thus, there is an urgent need for novel antibiotics that could be effective against B. anthracis. Antibacterials with a

Chart 1. Eremomycin and Vancomycin Adamantyl-Containing Amides

† Part of this work was presented at the annual 45th ICAAC 2005, Washington (U.S.A.). * To whom corresondence should be addressed. Tel.: 74952453753. Fax: 74952469980. E-mail: [email protected]. ‡ Anacor Pharmaceuticals, Inc. § Galileo Pharmaceuticals, Inc. | Gause Institute of New Antibiotics. ⊥ USAMRIID Bacteriology Division. # University of New Mexico.

10.1021/jm0700058 CCC: $37.00 © 2007 American Chemical Society Published on Web 07/03/2007

3682

Journal of Medicinal Chemistry, 2007, Vol. 50, No. 15

Maples et al.

Table 1. MICs for Adamantyl-Bearing Carboxamides of Glycopeptides 1a, 1b, 1c, 1d, and 2a, Comparing with Eremomycin (1), Vancomycin (2), and Teicoplanin against Staphylococci and Enterococci Clinical Isolates MIC (µg/mL) strain/isolate

eremomycin (1)

vancomycin (2)

teicoplanin

1a

1b

1c

1d

2a

Staphylococcus epidermidis 533 Staphylococcus hemeolyticus 602 Staphylococcus aureus 3793 (GISA) Staphylococcus aureus 3798 (GISA) Enterococcus faecium 568 (VSE) Enterococcus faecalis 559 (VSE) Enterococcus faecium 569 (GRE) Enterococcus faecalis 560 (GRE)

0.25 0.25 8 8 0.5 0.5 >128 >128

2 2 16 8 2 1 >128 >128

8 16 16 8 0.25 0.5 >128 >128

0.25 0.25 1 2 0.5 0.5 4 8

0.5 1 2 2 1 1 16 16

0.5 1 4 2 0.5 1 16 16

0.5 0.5 1 2 0.5 1 16 16

1 1 4 4 0.5 0.5 64 64

Table 2. Antibacterial Activity of 1a in Comparison with Vancomycin (2) MIC (µg/mL) strains/isolate

vancomycin (2)

1a

Staphylococcus aureus Stau_29213a Staphylococcus aureus Stau_33591a Staphylococcus aureus Stau_b11386a Staphylococcus aureus Stau_Mu50-HIP5406a Staphylococcus aureus Stau_HIP5827a Enterococcus faecalis Enfa_29212b Enterococcus faecalis Enfa_t29862c Enterococcus faecalis Enfa_vre-2c Streptococcus pyogenes Stpy_8668 Escherichia coli Esco_25922

1 1 1 2 8 2 >64 >64 0.5 >50

0.78 0.78 0.78 0.4 1.56 0.4 6.25 12.5 0.2 >50

c

a All staphylococcus strains are MRSA. b Vancomycin susceptible strain. Vancomycin resistant strains.

long half-life and deep tissue penetration are needed for effective therapy against multiple infectious agents, including B. anthracis. Thus semisynthetic antibiotics, derived from glycopeptides, may have better pharmacokinetic properties and offer therapeutic advantages. The glycopeptides (vancomycin 2 and teicoplanin) are traditionally the antibiotics of last choice for serious infections due to Gram-positive pathogens. Eremomycin (1, Chart 1) is a glycopeptide in the vancomycin group that is several times more active in vitro and in vivo than vancomycin,4 although not active against vancomycin-resistant Gram-positive organisms. The search for derivatives of glycopeptide antibiotics active against multidrug-resistant Gram-positive pathogens resulted in the discovery of derivatives of vancomycin, eremomycin, and other members of this group of antibiotics. Many of these compounds are active against methicillin-resistant S. aureus (MRSAa), glycopeptide intermediately resistant staphylococci (GISA), and vancomycin-resistant enterococci (GRE).5-7 In addition, a series of N′-substituted derivatives of eremomycin or vancomycin bearing hydrophobic moieties of the optimal size are generally active against glycopeptide-resistant enterococci (MICs ) 2-16 µg/mL) and act in a manner different from that of the parent antibiotics.8 It was demonstrated that nonbonded interactions of the hydrophobic substituent at the amide group of eremomycin amide and the L-Ala-D-iso-Glu component of the peptidoglycan chain results in steric hindrance in interaction with the bacterial transglycosylase enzyme.9 However, some hydrophobic glycopeptides amides, for example, decyl amides, are a Abbreviations: MIC, minimal inhibitory concentration of antibiotic (µg/ml); MSSA, methicillin-susceptible S. aureus; MRSA, methicillinresistant S. aureus; GISA, staphylococci with an intermediate resistance to vancomycin; GRE, vancomycin-resistant enterococci; DPPA, diphenylphosphoryl azide; PyBOP, benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate; HBPyU, O-(benzotriazol-1-yl)-1,1,3,3-bis(tetramethylene)uronium hexafluorophosphate; HBTU, O-(benzotriazol-1-yl)-1,1,3,3tetramethyluronium tetrafluoroborate; DIPEA, di-isopropylethylamine; ESI MS, electrospray ionization mass spectra.

Table 3. MIC of 1a and Ciprofloxacin against B. anthracis Strains and Isolates MICb (µg/mL)

B. anthracis strain/isolatea

1a

ciprofloxacin

Ames HH105-5 HH105-6 HH105-5R HHT105-5R HH113-6R

0.50 0.50 0.50 0.25 0.25 0.50

0.06 8.00 32.00 4.00 8.00 4.00

a The original B. anthracis Ames strain spore stock was obtained from the U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD. The resistant strains came from the studies using the Ames strain in animals, as described by Lyons et al.15 b The values for all strains were within a well (single dilution) of the parent Ames strain.

Table 4. Mortality and ED50 Estimates for 1a in a S. aureus Infection Model dose (mg/kg)

mortalities

ED50

lower 95% C.I.

upper 95% C.I.

1 5 10 25 50

5 4 4 3 0

20.4

9.8

42.5

cytotoxic.10 One approach to overcome the cytotoxicity of hydrophobic glycopeptide derivatives is the introduction of a hydrophilic moiety into the glycopeptide molecule to compensate for the effects of the hydrophobic substituent. This strategy led to telavancin, a derivative of vancomycin carrying simultaneously both hydrophilic and hydrophobic substituents.11 An alternative approach to minimize toxicity is to carefully select hydrophobic substituents that will exert only the desired effect on resistant strains and not cause any concomitant toxicological effects. Adamantane derivatives, such as amantadine, rimantadine, memantine, and some others, are broadly employed as antiviral or neurological drugs and demonstrate good pharmacological properties.12 Earlier we demonstrated that adamantyl-containing amides of vancomycin and eremomycin aglycons exhibit low cytotoxic effects and are active against staphylococci and enterococci clinical isolates in vitro (2-32 µg/mL).13 These results prompted us to synthesize and study a series of adamantyl-derived amides of fully glycosylated glycopeptides and to evaluate their antibacterial activity. Chemistry Earlier we described the synthesis of several eremomycin carboxamides by a one-step reaction of the antibiotic with the appropriate amine in the presence of a condensing reagent such as DPPA, PyBOP, HBPyU, or HBTU.8,14 Carboxamides of eremomycin bearing the adamantyl-moiety, 2-amino-adamantane 1a, 1-amino-adamantane 1b, 1-(adamantyl-1)ethylamine

Semisynthetic DeriVatiVe of Antibiotic Eremomycin

Journal of Medicinal Chemistry, 2007, Vol. 50, No. 15 3683

Figure 1. Initial dose-response study showing prevention by 1a of mortality in mice exposed to a lethal challenge of B. anthracis spores by intratracheal instillation.

1c, and (adamantyl-1)methylamine 1d and 2-amino-adamantyl amide of vancomycin (2a; Chart 1), were obtained by the condensation of 1 or 2 with the corresponding amines in the presence of PyBOP, HBPyU, or HBTU reagents in DMSO without protection of antibiotic amino groups. The yields of 1a-d and 2a were ∼60-70%, with HPLC purity at 95-97%. The homogeneity and identity of these compounds were assessed by two systems of HPLC and two systems of TLC, and electrospray ionization mass-spectrometry (ESI MS; Table in Supporting Information). Biological Evaluation Comparative in vitro antibacterial activities of adamantanederived amides of eremomycin and vancomycin against staphylococci and enterococci strains versus eremomycin, vancomycin, and teicoplanin are presented in Table 1. The four amides of eremomycin (1a-d) were found to be as active as the natural antibiotics against vancomycin-sensitive bacterial strains (MICs 0.25-0.5 µg/mL), more active than the natural antibiotics against GISA staphylococci (1-4 µg/mL), and only 1a was active (4-8 µg/mL) against vancomycin-resistant enterococci. In contrast to 1a, (adamantyl-2)-amide of vancomycin (2a) was less active against vancomycin-sensitive and especially vancomycin-resistant strains (64 µg/mL). Compound 1a was further evaluated in vitro against five Staphylococcus aureus strains, one Streptococcus pyogenes strain, and one vancomycinsensitive enterococci strain and showed high activity. It was active against two vancomycin-resistant Enterococcus faecalis strains (6.25-12.5 mcg/mL; Table 2). Antibiotic susceptibilities were also determined for the Ames strain of B. anthracis and five strains isolated from failed-ciprofloxacin-treated mice that had been challenged with the Ames strain. In vitro antibacterial activities of 1a (against B. anthracis strains) in comparison with ciprofloxacin are presented in Table 3. These strains represent a range of decreased susceptibility to ciprofloxacin in vitro. The susceptibility pattern for 1a was unaffected by the alteration in ciprofloxacin susceptibility. As shown in Table 3, the MICs for 1a for all strains range within a single dilution of the parent Ames strain (0.25-0.5 µg/mL). Cytotoxicity for 1a was IC50 > 200 µM (CEM0 cells, measured by Balzarini, J. et al. by the method described earlier).10 The objective of the pharmacokinetic study was to assess the exposure of female BALB/c mice to 1a when the test compound was administered by intravenous (iv), subcutaneous (sc), intraperitoneal (ip), and oral (po) routes. The bioavailability of 1a from sc, ip, and po dosing routes to mice was 78, 95, and 0%, respectively, and the Cmax (µg/mL) values from iv, sc, and ip dosing were 11.5, 7.2, and 9.7, respectively. The pharmacokinetic parameters of 1a iv were the following: half-life ) 2.8 h, clearance ) 536 mL/h/kg, and volume of distribution Vss )

2.18 L/kg. There was no mortality or significant clinical observations noted during the course of this study. The in vivo efficacy for 1a was demonstrated in several tests, including survival studies against Staphylococcus aureus (S. aureus) and B. anthracis (Ames strain). Compound 1a was evaluated for its ability to prevent mortality in mice caused with a lethal dose of S. aureus PGO#172 (ATCC#29213). Compound 1a was administered as an iv bolus at doses 1, 5, 10, 25, or 50 mg/kg to groups of five mice. As shown in Table 4, 1a provided dose-dependent efficacy with an ED50 of 20.4 mg/kg. With regard to B. anthracis, the efficacy of 1a was first tested against a systemic bacteremia in mice that were inoculated intravenously with a known number of vegetative bacilli. In this study, two single daily intravenous doses of either 25 or 50 mg/kg of compound 1a were sufficient for complete protection against a lethal B. anthracis challenge (data not shown). Ciprofloxacin at 106 mg/kg/day for 3 days also provided complete protection, as expected. Most impressively, 1a is equivalent or possibly slightly superior in efficacy to ciprofloxacin in preventing mortality in mice infected with lethal doses of B. anthracis. The efficacy of 1a was then examined in a pulmonary anthrax model in which mice were inoculated with B. anthracis (Ames) spores. Daily subcutaneous treatment with 10, 30, or 100 mg/ kg of 1a for 6 days provided significant protection against a lethal pulmonary challenge with B. anthracis spores, with 88% survival observed in the mice treated with 10 or 30 mg/kg and 100% survival in the mice treated with the 100 mg/kg dose (Figure 1). In comparison, oral treatment with ciprofloxacin at a daily dose of 135 mg/kg for 6 days resulted in 75% protection in this study. No increase in survival with 1a was observed by treating the mice with 30 mg/kg twice a day as compared to once a day (86% survival vs 88% survival, respectively; p ) 0.96). In a second study, the treatment dose of 1a was further reduced, and the efficacy again was tested against a pulmonary challenge with B. anthracis spores. As shown in Figure 2, a significant increase in survival was observed at all doses of 1a as compared to the vehicle-treated mice, and the protection was dose-dependent with 85.7, 62.5, 50, and 12.5% survival observed in the groups treated with 1a at doses of 10, 3, 1, or 0.3 mg/ kg/day, respectively, while 87.5% protection was observed in mice treated orally with Ciprofloxacin at 114 mg/kg. Discussion The 2-adamantyl amide of eremomycin 1a was selected from the series of adamantyl-derived amides of eremomycin and vancomycin as the best compound with antibacterial activity against 17 clinical isolates of Gram-positive bacteria. It was distinguished by having a 2.8 h half-life in mice and a volume

3684

Journal of Medicinal Chemistry, 2007, Vol. 50, No. 15

Maples et al.

Figure 2. Dose-response study of 1a showing prevention of mortality in mice exposed to a lethal challenge of B. anthracis spores by intratracheal instillation.

of distribution (Vss) of 2.18 L/kg. The Vss of 1a is much higher than that for marketed glycopeptides (700 mL/kg) and implies excellent deep tissue penetration. This property of 1a is important since antibacterials with long half-lives and deep tissue penetration may be beneficial for skin and soft tissue infections. Conclusion The adamantyl amide of eremomycin, 1a, had excellent activity against a range of clinical isolates, including MSSA, MRSA, and GISA, and moderate activity against GRE. In vivo efficacy for 1a was demonstrated by its ability to prevent death caused by either S. aureus or B. anthracis. Compound 1a is very effective at preventing mortality induced by B. anthracis, is significantly more efficacious than ciprofloxacin in this animal model, and is effective in vitro against B. anthracis strains shown to be resistant to ciprofloxacin. These compelling results are highly suggestive that 1a has considerable promise as a development candidate for use as a therapeutic agent for the treatment of a B. anthracis infection. Thus, 1a represents a promising development candidate for use as a therapeutic countermeasure against B. anthracis exposure. Further studies are needed to recognize the full potential of this novel compound. Experimental Section Chemistry. Synthesis of 1a. Compound 1a was synthesized by the condensation of antibiotic eremomycin sulfate (1 equiv) with (adamanthyl-2) amine hydrochloride (3 equiv) using PyBOP (1.1 equiv) in DMSO in the presence of DIPEA (pH ∼8) using TLC to monitor reaction progress with UV detection. After 20 min of stirring at 18 °C, 50 mL of Et2O was added to the reaction mixture and the mixture was shaken intensively to extract DMSO partly. The layer of Et2O was separated. The DMSO layer was poured into 200 mL of a stirring acetone to precipitate the product of the reaction, which was filtered off, washed with acetone, and dried in vacuum to give a white powder of crude 1a. Then crude 1a was dissolved in 9 mL of water and applied to a chromatographic column with silanized silica gel, pre-equilibrated with water (1 g of solid for 70 cm3 of silica gel). The column was eluted with water. Fractions containing the pure 1a were combined, acidified with 6 N H2SO4 to pH 2, and passed through a column with DOWEX 50WX2 resin (Serva, mesh size 200-400 µm) with 0.25 N NH4OH as eluent. The eluates were concentrated in vacuum to the volume ∼50 mL and adjusted to pH 7 with 1 N aqueous solution of H2SO4. The solution was evaporated to a volume of 15 mL and 400 mL of acetone was added to precipitate the product. The precipitate was filtered, washed with acetone, and dried in vacuum to give a white powder of 2-adamantylamide of eremomycin sulfate (1a) in a yield of 62%. HPLC purity was ∼97%. TLC, HPLC, and ESI MS data are presented in the Supporting Information (SI).

Synthesis of 1b, 1c, 1d, and 2a. The compounds 1b-d and 2a were obtained by the procedure similar for 1a. They were purified by column chromatography over silanized silica gel. The yields of 1b-d and 2a were ∼60-70%, HPLC purity was 95-97%. HPLC, TLC, and ESI MS data for adamantyl-bearing carboxamides of eremomycin (1b-d) and vancomycin (2a) are presented in the SI. Biological Evaluation. Antibiotic Susceptibilities against Staphylococci and Enterococci (Table 1 and 2). Minimum inhibitory concentrations (MICs) were determined by the microdilution method using Mueller Hinton broth, in 96-well plates, as recommended by NCCLS. Antibiotic stocks were diluted to 250 µg/mL with cation-adjusted Mueller-Hinton broth (CAMHB) and serially diluted 2-fold in 50 µL of CAMHB in the wells. The antibiotic range was 64 to 0.03 µg/mL based on a final well volume of 100 µL after inoculation. Mortality and ED50 for 1a in S. aureus Mouse Model. The mice were infected by S. aureus PGO#172 (ATCC#29213) strain by intraperitoneal injection and treated with 1a intravenously (Table 4). Experimental details for antibiotic susceptibility studies against B. anthracis (Table 3), pharmacokinetic studies in BALB/c mice, and mouse efficacy studies (Figures 1 and 2) are presented in the SI.

Acknowledgment. The authors thank M. I. Reznikova, Ph.D. for HPLC, E. P. Mirchink, Ph.D. M.D. for studying the antibacterial activity, and T. A. Loim, N. M. Malutina, and E. B. Isakova for excellent assistance (all from Gause Institute of New Antibiotics). Initial development of 1a was sponsored under a contract from the U.S. Department of Defense in an effort to identify and develop novel antibiotics for use against biowarfare pathogens. Supporting Information Available: Experimental for chemistry, pharmacokinetic study in BALB/c mice, and mouse efficacy studies. This material is available free of charge via the Internet at http//pubs.acs.org.

References (1) Brook, I. The prophylaxis and treatment of anthrax. Int. J. Antimicrob. Agents 2002, 20, 320-325. (2) Greenfield, R. A.; Bronze, M. S. Current therapy and the development of therapeutic options for the treatment of diseases due to bacterial agents of potential biowarfare and bioterrorism. Curr. Opin. InVest. Drugs 2004, 5, 135-140. (3) Athamna, A.; Athamna, M.; Abu-Rashed, N.; Medlej, B.; Bast, D. J.; Rubinshtein, E. Selection of Bacillus anthracis isolates resistant to antibiotics. J. Antimicrob. Chemother. 2004, 54, 424-428. (4) Gause, G. F.; Brazhnikova, M. G.; Lomakina, N. N.; Berdnikova, T. F.; Fedorova, G. B.; Tokareva, N. L.; Borisova, V. N.; Batta, G. Y. EremomycinsNew glycopeptide antibiotic: Chemical properties and structure. J. Antibiot. 1989, 42 (12), 1790-1799. (5) Malabarba, A.; Nicas, T. I.; Thompson, R. S. Structural modifications of glycopeptide antibiotics. Med. Res. ReV. 1997, 17, 69-137.

Semisynthetic DeriVatiVe of Antibiotic Eremomycin (6) Ge, M.; Chen, Z.; Onishi, H. R.; Kohler, J.; Silver, L. L.; Kerns, R.; Fukuzawa, S.; Thompson, C.; Kahne, D. Vancomycin derivatives that inhibit peptidoglycan biosynthesis without binding D-Ala-D-Ala. Science 1999, 284, 507-511. (7) Preobrazhenskaya, M. N.; Olsufyeva, E. N. Patents on glycopeptides of the vancomycin family and their derivatives as antimicrobials. Expert Opin. Ther. Pat. 2004, 14 (2), 141-173. (8) Printsevskaya, S. S.; Pavlov, A. Y.; Olsufyeva, E. N.; Mirchink, E. P.; Isakova, E. B.; Reznikova, M. I; Goldman, R. C.; Brandstrom, A. A.; Baizman, E. R.; Longley, C. B.; Sztaricskai, F.; Batta, G.; Preobrazhenskaya, M. N. Synthesis and mode of action of hydrophobic derivatives of glycopeptide antibiotic eremomycin and des(N-methyl-D-leucyl)eremomycin against glycopeptide-sensitive and -resistant bacteria. J. Med. Chem. 2002, 45, 1340-1347. (9) Kim, S. J.; Cegelski, L.; Preobrazhenskaya, M.; Schaefer, J. Structures of Staphylococcus aureus cell-wall complexes with vancomycin, eremomycin, and chloroeremomycin derivatives by 13C{19F} and 15N{19F} rotational-echo double resonance. Biochemistry 2006, 45, 5235-5250. (10) Balzarini, J.; Pannecouque, C.; DeClercq, E.; Pavlov, A. Y.; Printsevskaya, S. S.; Miroshnikova, O. V.; Reznikova, M. I.; Preobrazhenskaya, M. N. Antiretroviral activity of semisynthetic derivatives of glycopeptide antibiotics. J. Med. Chem. 2003, 46 (13), 2755-2764.

Journal of Medicinal Chemistry, 2007, Vol. 50, No. 15 3685 (11) Leadbetter, M. R.; Adams, S. M.; Bazzini, B.; Fatheree, P. R.; Karr, D. E.; Krause, K. M.; Lam, B. M. T.; Linsell, M. S.; Nodwell, M. B.; Pace, J. L.; Quast, K.; Shaw, J.-P.; Soriano, E.; Trapp, S. G.; Villena, J. D.; Wu, T. X.; Christensen, B. G.; Judice, J. K. Hydrophobic vancomycin derivatives with improved ADME properties: discovery of telvancin (TD-6424). J. Antibiot. 2004, 57 (5), 326-336. (12) Morosov, I. S.; Petrov, V. I.; Sergeeva, S. A. Pharmacology of Adamanatane; 2001; pp 50-60. (13) Printsevskaya, S. S; Solovieva, S. E.; Olsufyeva, E. N.; Mirchink, E. P.; Isakova, E. B.; De Clercq, E.; Balzarini, J.; Preobrazhenskaya, M. N. Structure-activity relationship studies of a series of antiviral and antibacterial aglycon derivatives of the glycopeptide antibiotics vancomycin, eremomycin, and dechloroeremomycin. J. Med. Chem. 2005, 48 (11), 3885-3890. (14) Miroshnikova, O. V.; Printsevskaya, S. S.; Olsufyeva, E. N.; Nilius, A.; Hensey-Rudloff, D.; Preobrazhenskaya, M. N. Structure-activity relationships in the series of eremomycin carboxamides. J. Antibiot. 2000, 53 (3), 286-293. (15) Lyons, C. R.; Lovchik, J.; Hutt, J.; Lipscomb, M. F.; Wang, E.; Heninger, S.; Berliba, L.; Garrison, K. Murine model of pulmonary anthrax: Kinetics of dissemination, histopathology, and mouse strain susceptibility. Infect. Immun. 2004, 72, 4801-4809.

JM0700058